Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Am J Physiol Lung Cell Mol Physiol ; 309(10): L1037-40, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26432873

ABSTRACT

Supplemental oxygen after premature birth results in aberrant airway, alveolar, and pulmonary vascular development with an increased risk for bronchopulmonary dysplasia, and development of wheeze and asthma, pulmonary hypertension, and chronic obstructive pulmonary disease in survivors. Although stimulation of the nitric oxide (NO)-soluble guanylate cyclase (sGC)-cGMP signal transduction pathway has significant beneficial effects on disease development in animal models, so far this could not be translated to the clinic. Oxidative stress reduces the NO-sGC-cGMP pathway by oxidizing heme-bound sGC, resulting in inactivation or degradation of sGC. Reduced sGC activity and/or expression is associated with pathology due to premature birth, oxidative stress-induced lung injury, including impaired alveolar maturation, smooth muscle cell (SMC) proliferation and contraction, impaired airway relaxation and vasodilation, inflammation, pulmonary hypertension, right ventricular hypertrophy, and an aggravated response toward hyperoxia-induced neonatal lung injury. Recently, Britt et al. (10) demonstrated that histamine-induced Ca(2+) responses were significantly elevated in hyperoxia-exposed fetal human airway SMCs compared with normoxic controls and that this hyperoxia-induced increase in the response was strongly reduced by NO-independent stimulation and activation of sGC. These recent studies highlight the therapeutic potential of sGC modulators in the treatment of preterm infants for respiratory distress with supplemental oxygen. Such treatment is aimed at improving aberrant alveolar and vascular development of the neonatal lung and preventing the development of wheezing and asthma in survivors of premature birth. In addition, these studies highlight the suitability of fetal human airway SMCs as a translational model for pathological airway changes in the neonate.


Subject(s)
Enzyme Activators/pharmacology , Guanylate Cyclase/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Respiratory Distress Syndrome, Newborn/drug therapy , Animals , Asthma/etiology , Enzyme Activators/therapeutic use , Humans , Molecular Targeted Therapy , Oxygen Inhalation Therapy/adverse effects , Respiratory Distress Syndrome, Newborn/enzymology , Signal Transduction , Soluble Guanylyl Cyclase
2.
Clin Respir J ; 9(4): 450-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-24796387

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the possible relationship between angiotensin-converting enzyme (ACE) gene polymorphism (D/D and I/D genotypes) and respiratory distress syndrome (RDS) in preterm neonates. STUDY DESIGN: Our study included 120 preterm neonates (<37 weeks of gestation) with RDS (the patient group) and 120 preterm neonates without RDS (the control group). Blood samples were obtained from patients and control groups, and ACE gene polymorphism was analysed using the polymerase chain reaction method. RESULTS: D/D genotype was highly significant in the patient group compared with the control group (48.3% of RDS group vs 20% of the control group, P < 0.001). Meanwhile, I/D and I/I genotypes were significantly higher in the control group (75% and 5% of the control group vs 50% and 1.7% of the patient group, P < 0.001). D/D genotype was highly significant in neonates with bronchopulmonary dysplasia (BPD) compared with I/D genotype (P = 0.001). CONCLUSION: Our results may suggest that D/D genotype is associated with increased risk of RDS and BPD development in preterm neonates.


Subject(s)
Infant, Premature/physiology , Peptidyl-Dipeptidase A/genetics , Respiratory Distress Syndrome, Newborn/genetics , Case-Control Studies , Female , Genetic Predisposition to Disease , Genotype , Humans , Infant, Newborn , Male , Polymorphism, Genetic , Respiratory Distress Syndrome, Newborn/enzymology , Retrospective Studies
3.
Pediatr Res ; 76(6): 515-21, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25188742

ABSTRACT

BACKGROUND: Adenosine levels are regulated by ecto-5'-nucleotidase/CD73 and adenosine deaminase (ADA). Adenosine regulates endothelial permeability and anti-inflammatory responses via adenosine receptors. Here, the adenosine receptors and purine-converting enzymes were studied during postnatal development and inflammation. METHODS: Newborn, 1-, 10-, 14-d-old and adult C57BL/6 mice were challenged intraperitoneally (i.p.) with lipopolysaccharide (LPS) for 6 h. The inflammatory response was evaluated by histochemistry. Expression levels of adenosine receptors (A1, A2A, A2B, and A3), CD73, and ADA were measured by quantitative reverse transcription polymerase chain reaction. A1 was studied by immunohistochemistry, and enzyme activities were analyzed by thin-layer chromatography. RESULTS: LPS caused respiratory distress in newborns within 24 h. LPS induced neutrophils at the basal stage and alveolar congestion. Low activity and expression of CD73 increased after birth. Expression of ADA after LPS increased 16-fold in adults and 2-fold in newborns (P < 0.05). A1 expression was high in newborns and increased after LPS (P < 0.05). A1 was localized to endothelial membranes. A2A decreased after LPS in newborns and increased in adults (P < 0.05). The expression of A3 increased in newborns and adults after LPS. CONCLUSION: Low pulmonary CD73 expression, LPS-induced suppression of A2A, LPS-induced increase of A1 expression, and severe respiratory distress were distinguishing responses in the newborns from those in the adults.


Subject(s)
5'-Nucleotidase/metabolism , Adenosine Deaminase/metabolism , Adenosine/metabolism , Lipopolysaccharides , Lung/enzymology , Pneumonia/enzymology , Receptors, Purinergic P1/metabolism , Respiratory Distress Syndrome, Newborn/enzymology , 5'-Nucleotidase/genetics , Adenosine Deaminase/genetics , Age Factors , Animals , Animals, Newborn , Disease Models, Animal , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Enzymologic , Lung/growth & development , Mice, Inbred C57BL , Neutrophil Infiltration , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/physiopathology , RNA, Messenger/metabolism , Receptors, Purinergic P1/genetics , Respiratory Distress Syndrome, Newborn/chemically induced , Respiratory Distress Syndrome, Newborn/genetics , Respiratory Distress Syndrome, Newborn/physiopathology
4.
Ital J Pediatr ; 40(1): 27, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24602444

ABSTRACT

AIM: To evaluate the association of NOS1 and NOS3 gene polymorphisms with the risk/severity of neonatal respiratory distress syndrome (RDS) among preterm infants. METHODS: The patient group was 189 preterm infants diagnosed with RDS. The control group was 227 preterm neonates who did not develop RDS. NOS genotyping was performed using an improved multiplex ligation detection reaction (iMLDR) technique based on LDR. RESULTS: It was found that genotype and allele frequencies of rs2682826 of the NOS1 gene and rs1799983 of the NOS3 gene were not significantly different between the RDS group and the control group. However, when the preterm infants were divided into two and three groups based on gestational age and birth weight, a study of the SNP rs1799983 of the NOS3 gene showed that the GG genotype and G allele frequencies were significantly increased in the RDS groups, the GT genotype and A allele were less frequent among the RDS groups in 26-32.9 weeks of gestational age and in a birth weight subgroup of <1.5 Kg. CONCLUSION: Our study raises the possibility that a genetic variation of NOS3 could be implicated in the pathophysiology of RDS in the Chinese Han population, especially in very preterm and very low birth weight infants.


Subject(s)
DNA/genetics , Genetic Predisposition to Disease , Infant, Premature, Diseases/genetics , Infant, Premature , Nitric Oxide Synthase/genetics , Polymorphism, Genetic , Respiratory Distress Syndrome, Newborn/genetics , Adult , Alleles , China/epidemiology , Female , Gene Frequency , Genotype , Gestational Age , Humans , Incidence , Infant, Newborn , Infant, Premature, Diseases/enzymology , Infant, Premature, Diseases/epidemiology , Male , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Polymerase Chain Reaction , Respiratory Distress Syndrome, Newborn/enzymology , Respiratory Distress Syndrome, Newborn/epidemiology
5.
Pediatr Crit Care Med ; 14(9): e438-41, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24226567

ABSTRACT

OBJECTIVE: Angiotensin-converting enzyme and its effector peptide angiotensin II have been implicated in the pathogenesis of acute respiratory distress syndrome. Recently, angiotensin-converting enzyme 2 was identified as the counter-regulatory enzyme of angiotensin-converting enzyme that converts angiotensin II into angiotensin-(1-7). The aim of this study was to determine pulmonary angiotensin-converting enzyme and angiotensin-converting enzyme 2 activity in patients with acute respiratory distress syndrome. DESIGN: Prospective observational pilot study. SETTING: A PICU of a university hospital. PATIENTS: Fourteen patients admitted, requiring mechanical ventilation for respiratory syncytial virus lower respiratory tract infection. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: Two groups of patients were distinguished at admission: a group fulfilling the criteria for acute respiratory distress syndrome and a non-acute respiratory distress syndrome group. Angiotensin-converting enzyme and angiotensin-converting enzyme 2 activity were measured in bronchoalveolar lavage fluid. Patients with acute respiratory distress syndrome had increased angiotensin-converting enzyme activity and decreased angiotensin-converting enzyme 2 activity (p < 0.001) compared with the control group. CONCLUSION: It is shown for the first time that in acute respiratory distress syndrome, enhanced angiotensin-converting enzyme activity is paralleled by a reduced angiotensin-converting enzyme 2 activity, similar to that found in an experimental rat model of acute respiratory distress syndrome. The reduced angiotensin-converting enzyme 2 activity may be counteracted by restoring angiotensin-(1-7) level, thereby offering a novel treatment modality for this syndrome.


Subject(s)
Peptidyl-Dipeptidase A/metabolism , Respiratory Distress Syndrome, Newborn/enzymology , Angiotensin-Converting Enzyme 2 , Bronchoalveolar Lavage Fluid/chemistry , Case-Control Studies , Female , Humans , Infant , Infant, Newborn , Lung/enzymology , Male , Peptidyl-Dipeptidase A/analysis , Prospective Studies
6.
Crit Care ; 17(4): R163, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23883784

ABSTRACT

INTRODUCTION: Secretory phospholipase A2 is supposed to play a role in acute lung injury but no data are available for pediatric acute respiratory distress syndrome (ARDS). It is not clear which enzyme subtypes are secreted and what the relationships are between enzyme activity, biophysical and biochemical parameters, and clinical outcomes. We aimed to measure the enzyme and identify its subtypes and to study its biochemical and biophysical effect. The secondary aim was to correlate enzyme activity with clinical outcome. METHODS: Bronchoalveolar lavage was performed in 24 infants with ARDS and 14 controls with no lung disease. Samples were assayed for secretory phospholipase A2 and molecules related to its activity and expression. Western blotting and captive bubble surfactometry were also performed. Clinical data were real time downloaded. RESULTS: Tumor necrosis factor-α (814 (506-2,499) vs. 287 (111-1,315) pg/mL; P = 0.04), enzyme activity (430 (253-600) vs. 149 (61-387) IU/mL; P = 0.01), free fatty acids (4.3 (2.8-8.6) vs. 2 (0.8-4.6) mM; P = 0.026), and minimum surface tension (25.6 ± 6.1 vs. 18 ± 1.8 mN/m; P = 0.006) were higher in ARDS than in controls. Phospholipids are lower in ARDS than in controls (76.5 (54-100) vs. 1,094 (536-2,907) µg/mL; P = 0.0001). Three enzyme subtypes were identified (-IIA, -V, -X), although in lower quantities in controls; another subtype (-IB) was mainly detected in ARDS. Significant correlations exist between enzyme activity, free fatty acids (ρ = 0.823; P < 0.001), and surface tension (ρ = 0.55; P < 0.028). Correlations also exist with intensive care stay (ρ = 0.54; P = 0.001), PRISM-III24 (ρ = 0.79; P< 0.001), duration of ventilation (ρ = 0.53; P = 0.002), and oxygen therapy (ρ = 0.54; P = 0.001). CONCLUSIONS: Secretory phospholipase A2 activity is raised in pediatric ARDS and constituted of four subtypes. Enzyme correlates with some inflammatory mediators, surface tension, and major clinical outcomes. Secretory phospholipase A2 may be a clinically relevant target in pediatric ARDS.


Subject(s)
Phospholipases A2, Secretory/physiology , Respiratory Distress Syndrome, Newborn/enzymology , Bronchoalveolar Lavage/methods , Enzyme Activation/physiology , Female , Humans , Infant , Male , Respiration, Artificial/methods , Surface Tension
7.
PLoS One ; 7(10): e47066, 2012.
Article in English | MEDLINE | ID: mdl-23071714

ABSTRACT

BACKGROUND: Secretory phospholipase A2 (sPLA2) plays a pivotal role in acute respiratory distress syndrome (ARDS). This enzyme seems an interesting target to reduce surfactant catabolism and lung tissue inflammation. Varespladib is a specifically designed indolic sPLA2 inhibitor, which has shown promising results in animals and adults. No specific data in pediatric ARDS patients are yet available. METHODS: We studied varespladib in broncho-alveolar lavage (BAL) fluids obtained ex vivo from pediatric ARDS patients. Clinical data and worst gas exchange values during the ARDS course were recorded. Samples were treated with saline or 10-40-100 µM varespladib and incubated at 37°C. Total sPLA2 activity was measured by non-radioactive method. BAL samples were subjected to western blotting to identify the main sPLA isotypes with different sensitivity to varespladib. Results was corrected for lavage dilution using the serum-to-BAL urea ratio and for varespladib absorbance. RESULTS: Varespladib reduces sPLA2 activity (p<0.0001) at 10,40 and 100 µM; both sPLA2 activity reduction and its ratio to total proteins significantly raise with increasing varespladib concentrations (p<0.001). IC(50) was 80 µM. Western blotting revealed the presence of sPLA2-IIA and -IB isotypes in BAL samples. Significant correlations exist between the sPLA2 activity reduction/proteins ratio and PaO(2) (rho = 0.63;p<0.001), PaO(2)/FiO(2) (rho = 0.7; p<0.001), oxygenation (rho = -0.6; p<0.001) and ventilation (rho = -0.4;p = 0.038) indexes. CONCLUSIONS: Varespladib significantly inhibits sPLA2 in BAL of infants affected by post-neonatal ARDS. Inhibition seems to be inversely related to the severity of gas exchange impairment.


Subject(s)
Acetates/pharmacology , Bronchoalveolar Lavage Fluid , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Phospholipases A2, Secretory/antagonists & inhibitors , Phospholipases A2, Secretory/metabolism , Respiratory Distress Syndrome, Newborn/enzymology , Dose-Response Relationship, Drug , Humans , Infant , Infant, Newborn , Keto Acids
8.
Am J Obstet Gynecol ; 206(5): 447.e17-24, 2012 May.
Article in English | MEDLINE | ID: mdl-22445700

ABSTRACT

OBJECTIVE: To determine the impact of maternal and fetal single nucleotide polymorphisms in key betamethasone pathways on neonatal outcomes. STUDY DESIGN: DNA was obtained from women given betamethasone and their infants. Samples were genotyped for 73 exploratory drug metabolism and glucocorticoid pathway single nucleotide polymorphisms. Clinical variables and neonatal outcomes were obtained. Logistic regression analysis using relevant clinical variables and genotypes to model for associations with neonatal respiratory distress syndrome was performed. RESULTS: One hundred nine women delivering 117 infants were analyzed. Sixty-four infants (49%) developed respiratory distress syndrome. Multivariable analysis revealed that respiratory distress syndrome was associated with maternal single nucleotide polymorphisms in CYP3A5 (odds ratio [OR], 1.63; 95% confidence interval [CI], 1.16-2.30) and the glucocorticoid resistance (OR, 0.28; 95% CI, 0.08-0.95) and fetal single nucleotide polymorphisms in ADCY9 (OR, 0.17; 95% CI, 0.03-0.80) and CYP3A7*1E (rs28451617; OR, 23.68; 95% CI, 1.33-420.6). CONCLUSION: Maternal and fetal genotypes are independently associated with neonatal respiratory distress syndrome after treatment with betamethasone for preterm labor.


Subject(s)
Betamethasone/metabolism , Genetic Markers , Glucocorticoids/metabolism , Obstetric Labor, Premature , Polymorphism, Single Nucleotide , Respiratory Distress Syndrome, Newborn/prevention & control , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Adenylyl Cyclases/genetics , Adult , Arylsulfotransferase/genetics , Betamethasone/therapeutic use , Cytochrome P-450 CYP3A/genetics , Female , Genotyping Techniques , Glucocorticoids/therapeutic use , Humans , Infant, Newborn , Logistic Models , Multivariate Analysis , Pregnancy , ROC Curve , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Glucocorticoid/genetics , Respiratory Distress Syndrome, Newborn/enzymology , Respiratory Distress Syndrome, Newborn/genetics , Treatment Outcome
9.
J Clin Pharmacol ; 52(5): 729-37, 2012 May.
Article in English | MEDLINE | ID: mdl-21602519

ABSTRACT

Secretory phospholipase A2 (sPLA2), which links surfactant catabolism and lung inflammation, is associated with lung stiffness, surfactant dysfunction, and degree of respiratory support in acute respiratory distress syndrome and in some forms of neonatal lung injury. Varespladib potently inhibits sPLA2 in animal models. The authors investigate varespladib ex vivo efficacy in different forms of neonatal lung injury. Bronchoalveolar lavage fluid was obtained from 40 neonates affected by hyaline membrane disease, infections, or meconium aspiration and divided in 4 aliquots added with increasing varespladib or saline. sPLA2 activity, proteins, and albumin were measured. Dilution was corrected with the urea ratio. Varespladib was also tested in vitro against pancreatic sPLA2 mixed with different albumin concentration. Varespladib was able to inhibit sPLA2 in the types of neonatal lung injury investigated. sPLA2 activity was reduced in hyaline membrane disease (P < .0001), infections (P = .003), and meconium aspiration (P = .04) using 40 µM varespladib; 10 µM was able to lower enzyme activity (P = .001), with an IC(50) of 87 µM. An inverse relationship existed between protein level and activity reduction (r = 0.5; P = .029). The activity reduction/protein ratio tended to be higher in hyaline membrane disease. Varespladib efficacy was higher in vitro than in lavage fluids obtained from neonates (P < .001).


Subject(s)
Acetates/pharmacology , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Lung Injury/enzymology , Phospholipases A2, Secretory/antagonists & inhibitors , Albumins/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Female , Gestational Age , Humans , Hyaline Membrane Disease/enzymology , Infant, Newborn , Italy , Keto Acids , Male , Meconium Aspiration Syndrome/enzymology , Pancreas/enzymology , Phospholipases A2, Secretory/metabolism , Respiratory Distress Syndrome, Newborn/enzymology , Respiratory Tract Infections/enzymology
10.
Mol Cell Biol ; 31(5): 1054-65, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21189286

ABSTRACT

Pregnancy in women with diabetes is associated with a higher risk of perinatal complications. In particular, infants of diabetic mothers frequently suffer from respiratory distress syndrome (RDS), which is a leading cause of death in preterm infants and is considered to be primarily due to hyperinsulinemia in infants in response to maternal hyperglycemia. To elucidate the mechanism of how insulin signaling induces RDS, bronchoalveolar epithelium-specific Akt1 transgenic (TG) mice were generated. Akt1 overexpression in fetal lung epithelium resulted in RDS in preterm infants born by Caesarean section at embryonic day 18.5 (E18.5). The expression levels of hypoxia-inducible factor 2α (HIF-2α) and its target vascular endothelial growth factor (VEGF) were downregulated in the lung of Akt1 TG mice. Inhibition of the Akt-mammalian target of rapamycin (mTOR) signaling axis by rapamycin restored the expression of VEGF and improved the lung pathology of Akt1 TG pups. Rapamycin also attenuated the RDS phenotype in wild-type mice delivered preterm at E17.5. In cultured lung epithelial cells, insulin reduced VEGF expression and transcriptional activity of HIF-2 on VEGF promoter in an mTOR-dependent manner. Thus, aberrant activation of the Akt-mTOR pathway in lung epithelium plays a causal role in the pathogenesis of infant RDS, presumably through downregulation of HIF-2-dependent VEGF expression in the lung.


Subject(s)
Proto-Oncogene Proteins c-akt/metabolism , Respiratory Distress Syndrome, Newborn/enzymology , TOR Serine-Threonine Kinases/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Disease Models, Animal , Female , Humans , Infant, Newborn , Insulin/metabolism , Lung/enzymology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Proto-Oncogene Proteins c-akt/genetics , Respiratory Mucosa/enzymology , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/genetics , Vascular Endothelial Growth Factors/genetics , Vascular Endothelial Growth Factors/metabolism
11.
J Matern Fetal Neonatal Med ; 24(5): 703-7, 2011 May.
Article in English | MEDLINE | ID: mdl-20836741

ABSTRACT

Adenosine is produced in the inflammed and damaged lung where it plays roles in the regulation of inflammation and tissue remodeling. Adenosine deaminase (ADA) is an enzyme responsible for the degradation of adenosine. Our aim was to compare the levels of ADA between infants with and without respiratory distress syndrome (RDS) and to determine the relationship between plasma ADA levels and bronchopulmonary dysplasia (BPD). One-hundred and twenty-five premature infants who were admitted to our neonatal intensive care unit were included in the study. Eighty-one of these infants with RDS were study group and the other 44 infants without RDS served as controls. Blood collection was made in the first day of life at the end of 24th-h and was used for laboratory testing. In the RDS group, mean ADA level was 25.5 (± 4.5) U/l, and in controls it was 26.3 (± 5.7) U/l. There was no statistically significant difference (p = 0.326) in these groups although there was a statistically difference of ADA levels between BPD (34.5 ± 5.2 U/l) and non-BPD (24.6 ± 4.1) patients (p = 0.001). There was also a positive relationship between ADA levels and severity of BPD (r = + 0.845, p = 0.01). Perinatal inflammation is the key mechanism of BPD. ADA level in early postnatal life is elevated in infants with BPD and may be related with perinatal inflammation.


Subject(s)
Adenosine Deaminase/blood , Bronchopulmonary Dysplasia/enzymology , Respiratory Distress Syndrome, Newborn/enzymology , Bronchopulmonary Dysplasia/immunology , Humans , Infant, Newborn , Infant, Premature , Lymphocytes/physiology , Respiratory Distress Syndrome, Newborn/immunology
12.
Respir Res ; 11: 57, 2010 May 12.
Article in English | MEDLINE | ID: mdl-20462446

ABSTRACT

BACKGROUND: Angiotensin converting enzyme (ACE) gene contains a polymorphism, consisting of either the presence (I) or absence (D) of a 287 base pair fragment. Deletion (D) is associated with increased circulating ACE (cACE) activity. It has been suggested that the D-allele of ACE genotype is associated with power-oriented performance and that cACE activity is correlated with muscle strength. Respiratory muscle function may be similarly influenced. Respiratory muscle strength in infants can be assessed specifically by measurement of the maximum inspiratory pressure during crying (Pimax). Pressure-time index of the respiratory muscles (PTImus) is a non-invasive method, which assesses the load to capacity ratio of the respiratory muscles.The objective of this study was to determine whether increased cACE activity in infants could be related to greater respiratory muscle strength and to investigate the potential association of cACE with PTImus measurements as well as the association of ACE genotypes with cACE activity and respiratory muscle strength in this population. METHODS: Serum ACE activity was assayed by using a UV-kinetic method. ACE genotyping was performed by polymerase chain reaction amplification, using DNA from peripheral blood. PTImus was calculated as (Pimean/Pimax) x (Ti/Ttot), where Pimean was the mean inspiratory pressure estimated from airway pressure, generated 100 milliseconds after an occlusion (P0.1), Pimax was the maximum inspiratory pressure and Ti/Ttot was the ratio of the inspiratory time to the total respiratory cycle time. Pimax was the largest pressure generated during brief airway occlusions performed at the end of a spontaneous crying effort. RESULTS: A hundred and ten infants were studied. Infants with D/D genotype had significantly higher serum ACE activity than infants with I/I or I/D genotypes. cACE activity was significantly related to Pimax and inversely related to PTImus. No association between ACE genotypes and Pdimax measurements was found. CONCLUSIONS: These results suggest that a relation in cACE activity and respiratory muscle function may exist in infants. In addition, an association between ACE genotypes and cACE activity, but not respiratory muscle strength, was demonstrated.


Subject(s)
Crying , Inhalation , Meconium Aspiration Syndrome/enzymology , Muscle Strength , Peptidyl-Dipeptidase A/blood , Respiratory Distress Syndrome, Newborn/enzymology , Respiratory Muscles/physiopathology , Sleep Wake Disorders/enzymology , Biomarkers/blood , Female , Genotype , Greece , Humans , Infant , Infant, Newborn , Male , Meconium Aspiration Syndrome/genetics , Meconium Aspiration Syndrome/physiopathology , Peptidyl-Dipeptidase A/genetics , Phenotype , Pressure , Respiration, Artificial , Respiratory Distress Syndrome, Newborn/genetics , Respiratory Distress Syndrome, Newborn/physiopathology , Sleep Wake Disorders/genetics , Sleep Wake Disorders/physiopathology , Time Factors , Up-Regulation
13.
Neonatology ; 97(1): 1-9, 2010.
Article in English | MEDLINE | ID: mdl-19478531

ABSTRACT

BACKGROUND: The massive pulmonary neutrophil influx in respiratory distress syndrome (RDS) in preterm infants has been ascribed to the effect of leukotriene B(4) (LTB(4)). OBJECTIVES: To investigate whether secretory phospholipase A(2) (sPLA(2)), the rate-limiting enzyme in LTB(4) production, is present in lungs of RDS infants and stimulates neutrophil migration. METHODS: sPLA(2) was measured in tracheal aspirates from 15 preterm infants with RDS. The effect of aspirates on cord blood neutrophil migration was first measured, and the contribution of sPLA(2) was assessed by addition of its endogenous inhibitor Clara cell protein (CC16) or absorption of sPLA(2) from the aspirates. The role of intracellular signal transduction activation and LTB(4) formation in sPLA(2)-induced neutrophil migration was determined using purified sPLA(2), several inhibitors of signal transduction, a LTB(4) synthesis inhibitor and a LTB(4) receptor antagonist. RESULTS: All aspirates contained sPLA(2), which significantly stimulated neutrophil migration. Addition of CC16 or absorption of sPLA(2) abolished the stimulatory effect. All inhibitors significantly reduced sPLA(2)-induced neutrophil migration. CONCLUSIONS: sPLA(2) is present in tracheal aspirates of preterm infants with RDS. Human recombinant sPLA(2) and pancreatic type sPLA(2) stimulate in vitro cord blood neutrophil migration via activation of intracellular signal transduction pathways, LTB(4) production and receptor binding. We speculate that sPLA(2) contributes to pulmonary neutrophil influx in RDS. Further studies are needed to determine the potential of sPLA(2) inhibition as a treatment for RDS.


Subject(s)
Neutrophils/physiology , Phospholipases A2, Secretory/metabolism , Phospholipases A2, Secretory/pharmacology , Respiratory Distress Syndrome, Newborn/enzymology , Bronchoalveolar Lavage Fluid/chemistry , Cell Migration Assays, Leukocyte , Cell Movement/drug effects , Cells, Cultured , Female , Gestational Age , Humans , Infant, Newborn , Infant, Premature , Male , Respiratory Distress Syndrome, Newborn/pathology , Uteroglobin/pharmacology
14.
Bosn J Basic Med Sci ; 9(4): 335-41, 2009 Nov.
Article in English | MEDLINE | ID: mdl-20002001

ABSTRACT

Volume density of respiratory organs was studied in vitro in newborn babies at different age of gestation (abort, immature, premature and mature) using stereometric method. The total of 23 cases was subject to this study. The respiratory organs (trachea, lungs) were taken from autopsies of newborn babies exited from different causes. For this purpose the tissues were fixed in formalin (10%) solution, cut serially in 7micro and 10micro slabs. Volume density of the respiratory system was assessed stereometricaly using Universal testing system Weibel M 42. We observed that volume density of epithelia, musculature and glands were proportionally present in the tracheal tissue. Cellular interstitial tissue is consistently increasing and corresponds to the developmental stages of the newborn babies. The density of tracheal ganglions is greater in premature ages of immature and premature newborns (p<0,05). Decreased number of ganglion cells is observed in mature ages (p<0,05). This is caused by intensive ramification of ganglions from serosa to deeper layers of trachea right to epithelium. Medium diameter of tracheal ganglions is greater in mature newborn babies and corresponds to developmental ages of babies.


Subject(s)
Ganglion Cysts/pathology , Infant, Newborn , Respiratory Distress Syndrome, Newborn/pathology , Trachea/innervation , Acetylcholinesterase/metabolism , Choline O-Acetyltransferase/metabolism , Ganglion Cysts/enzymology , Humans , Infant, Premature , Lung/enzymology , Lung/innervation , Lung/pathology , Respiratory Distress Syndrome, Newborn/enzymology , Trachea/enzymology , Trachea/pathology
15.
Eur J Pediatr ; 167(12): 1379-83, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18301921

ABSTRACT

Oxidative stress is known to play an important role in the pathogenesis of certain severe illnesses in preterm infants. The enzyme heme oxygenase-1 (HO-1) participates in cytoprotection against oxygen radical injury. We have previously described the role of HO-1 in physiologic adaptation by demonstrating the induction of HO-1 in healthy mature neonates and asymptomatic preterm infants. Our current aim was to investigate the HO-1 expression in preterm infants with respiratory distress syndrome (RDS). We collected venous blood samples from 28 preterm infants with RDS on the 1st, 3rd and 5th days after birth. The HO-1 mRNA expression was determined by means of a competitive reverse transcriptase PCR technique, and a quantitative blood count was performed on the residual blood sample. A significant increase in HO-1 expression was found in the preterm infants with RDS as compared with both the healthy mature and the asymptomatic premature groups. The elevation was approximately eight-fold. The platelet count displayed a significant negative association with the HO-1 expression, and in the RDS prematures with thrombocytopenia the HO-1 induction was significantly greater than in those with a normal platelet count. In conclusion, the RDS of prematures is accompanied by an elevated HO-1 expression during the first 5 days of life, consistent with the inflammatory and oxidative characteristics of the disease.


Subject(s)
Heme Oxygenase-1/blood , Infant, Premature , Respiratory Distress Syndrome, Newborn/enzymology , Biomarkers/blood , Case-Control Studies , Female , Gene Expression , Gene Expression Regulation, Enzymologic , Heme Oxygenase-1/genetics , Humans , Infant, Newborn , Male , Pilot Projects , Polymerase Chain Reaction , RNA, Messenger/blood , Respiratory Distress Syndrome, Newborn/blood , Respiratory Distress Syndrome, Newborn/genetics , Reverse Transcriptase Polymerase Chain Reaction
16.
Am J Physiol Lung Cell Mol Physiol ; 293(1): L142-50, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17400600

ABSTRACT

Our previous work has shown that adult mice with overexpression of IL-6 and IL-13 in the lung have enhanced survival in hyperoxia associated with reduced hyperoxia-induced lung injury and cell death. We hypothesized that there are developmental differences in these responses in the adult vs. the newborn (NB) animal, and these responses have clinical relevance in the human NB. We compared the responses to 100% O(2) of NB IL-6 and IL-13 transgenic mice with wild-type littermate controls by evaluating mortality, lung tissue TUNEL staining, and mRNA expression using RT-PCR. We used ELISA to measure IL-6 levels in tracheal aspirates from human neonates. Our results show that, in contrast to the cytoprotective effects in mature mice, IL-6 caused significantly increased mortality, DNA injury, caspases, cell death regulator and angiogenic factor expression in hyperoxia in the NB. Furthermore, tracheal aspirate levels of IL-6 were significantly increased in premature neonates with respiratory distress syndrome who had an adverse outcome (bronchopulmonary dysplasia/death). In contrast to the protective effects in adults, there was no survival advantage to the NB IL-13 mice in hyperoxia. These findings imply that caution should be exercised in extrapolating results from the adult to the NB.


Subject(s)
Hyperoxia/metabolism , Hyperoxia/pathology , Interleukin-13/metabolism , Interleukin-6/metabolism , Lung/growth & development , Lung/pathology , Angiogenesis Inducing Agents/metabolism , Angiopoietin-2/genetics , Angiopoietin-2/metabolism , Animals , Animals, Newborn , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Caspase 3/metabolism , DNA/metabolism , Gene Expression Regulation , Humans , Infant, Newborn , Mice , Mice, Inbred C57BL , Mice, Transgenic , Respiratory Distress Syndrome, Newborn/enzymology , Survival Analysis
17.
Pediatr Crit Care Med ; 8(1): 40-6, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17149150

ABSTRACT

OBJECTIVE: To test the hypothesis that recombinant Clara cell secretory protein (rhCC10) instillation would foster improved lung function, acute structural preservation, and attenuation of matrix metalloproteinase (MMP) activity in a surfactant-deficient, mechanically ventilated lung. DESIGN: Interventional laboratory study. SETTING: An academic medical research facility in the northeastern United States. SUBJECTS: Sedated, ventilated premature lambs. INTERVENTIONS: Preterm lambs (n = 18; 126 +/- 3 days gestation) were instrumented, ventilated, and treated with 100 mg/kg exogenous surfactant. Lambs were randomized to receive 0, 0.5, or 5.0 mg/kg rhCC10 (n = 6 per group) and were ventilated for 4 hrs. MEASUREMENTS AND MAIN RESULTS: Posttreatment, lung function and cardiopulmonary stability were monitored for the ventilation period and then animals were killed for in vitro surfactant function analysis, lung histomorphometry, and analysis of MMP-2, -7, and -9 as well as their tissue inhibitors (TIMP)-1 and -2. Ventilation efficiency and pulmonary compliance were improved in the 5.0-mg/kg rhCC10 group by 4 hrs. Lung expansion was variable in the apical regions only. MMP-2 quantity was greater in the apical than the base lung regions of rhCC10-treated groups, and rhCC10 decreased MMP-7 in the base of the lung. CONCLUSIONS: These data suggest that improved lung function in the surfactant-treated preterm lamb following intratracheal rhCC10 may be related to the reduction of proteolytic activity of MMP-7.


Subject(s)
Enzyme Inhibitors/therapeutic use , Metalloproteases/metabolism , Respiration, Artificial , Respiratory Distress Syndrome, Newborn/therapy , Uteroglobin/therapeutic use , Animals , Animals, Newborn , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Humans , Infant, Newborn , Lung/pathology , Lung Compliance , Metalloproteases/antagonists & inhibitors , Protease Inhibitors/analysis , Pulmonary Alveoli/pathology , Pulmonary Surfactants/therapeutic use , Respiratory Distress Syndrome, Newborn/drug therapy , Respiratory Distress Syndrome, Newborn/enzymology , Respiratory Distress Syndrome, Newborn/pathology , Sheep , Time Factors , Tissue Inhibitor of Metalloproteinase-1/analysis , Tissue Inhibitor of Metalloproteinase-2/analysis , Treatment Outcome , Uteroglobin/administration & dosage
18.
Biol Neonate ; 89(1): 6-14, 2006.
Article in English | MEDLINE | ID: mdl-16155381

ABSTRACT

AIM: Matrix metalloproteinases (MMPs) play an eminent role in airway injury and remodelling. We explored the hypothesis that pulmonary MMP levels would differ early after birth (2-4 days) between infants with resolving respiratory distress syndrome (RDS) and infants developing chronic lung disease of prematurity (CLD). METHODS: Thirty-two prematurely born infants (gestational age < or =30 weeks) diagnosed with RDS were included. In 13 infants RDS resolved while 19 developed CLD. MMP-2 and MMP-9 in bronchoalveolar lavage (BAL) fluids collected on postnatal days 2, 4, 7 and 10 were analyzed by zymography and densitometry. Immunochemistry was performed on BAL cells and lung tissue to identify cellular sources of MMP-9 in RDS and CLD. RESULTS: Median MMP-9 levels increased significantly on day 2 in BAL fluid from patients with resolving RDS (median values MMP-9 = 42.0 arbitrary units (AU)) compared to CLD patients (MMP-9 = 5.4 AU). MMP-9 and neutrophil lipocalin-associated MMP-9 (NGAL) were significantly higher on day 4 in BAL fluid from resolving RDS (MMP-9 = 65.8 AU; NGAL = 16.1 AU) compared to CLD (MMP-9 = 25.4 AU; NGAL = 2.0 AU), Levels of MMP-9 and NGAL increased subsequently on days 7 and 10 in CLD. No differences in MMP-2 levels were detected between RDS and CLD. Neutrophils, macrophages and alveolar type-II epithelial cells were identified as potential sources of MMP-9. CONCLUSION: Our findings indicate differences in early MMP-9 BAL fluid levels between resolving RDS and developing CLD, which may relate to the ability to raise an early and adequate response to the initial injury.


Subject(s)
Matrix Metalloproteinase 9/analysis , Respiratory Distress Syndrome, Newborn/enzymology , Acute-Phase Proteins/analysis , Aging , Bronchoalveolar Lavage Fluid , Chronic Disease , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Gestational Age , Humans , Immunohistochemistry , Infant, Newborn , Infant, Premature, Diseases/enzymology , Intensive Care, Neonatal , Lipocalin-2 , Lipocalins , Lung/enzymology , Lung Diseases/enzymology , Lung Diseases/etiology , Matrix Metalloproteinase 2/analysis , Proto-Oncogene Proteins/analysis
19.
J Histochem Cytochem ; 52(9): 1231-40, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15314090

ABSTRACT

We studied cell-specific protein expression of all the major antioxidant enzymes (AOEs) and related proteins, such as copper-zinc superoxide dismutase (CuZnSOD), manganese SOD (MnSOD), extracellular SOD (ECSOD), catalase, the heavy and light chains of gamma-glutamylcysteine synthetase (gamma-GCS-l and gamma-GCS-h, also called glutamate cysteine ligase), the rate-limiting enzyme in glutathione synthesis, hemeoxygenase-1 (HO-1), and thioredoxin (Trx), in developing human lung, respiratory distress syndrome, and bronchopulmonary dysplasia by immunohistochemistry. Generally, after 17 weeks of gestational age, MnSOD was predominantly expressed in bronchial epithelium, alveolar epithelium, and macrophages, CuZnSOD was expressed in bronchial epithelium, ECSOD was expressed in bronchial epithelium, vascular endothelium, and the extracellular matrix, catalase was expressed in bronchial epithelium and alveolar macrophages, gamma-GCS-h was expressed in bronchial epithelium and endothelium, and gamma-GCS-l was expressed in bronchial epithelium. Trx was restricted to bronchial epithelium and to a lesser extent to alveolar macrophages, and HO-1 found in alveolar macrophages. Basically, the expression of these enzymes was similar in normal and diseased lung. It can be concluded that various AOEs and related proteins differ in their distribution and expression in lung before term, but generally it seems that infants are better adapted to high oxygen tension than might be expected.


Subject(s)
Antioxidants/metabolism , Bronchopulmonary Dysplasia/embryology , Bronchopulmonary Dysplasia/enzymology , Lung/embryology , Lung/enzymology , Respiratory Distress Syndrome, Newborn/embryology , Respiratory Distress Syndrome, Newborn/enzymology , Bronchopulmonary Dysplasia/pathology , Catalase/metabolism , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Glutamate-Cysteine Ligase/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1 , Humans , Infant , Infant, Newborn , Lung/pathology , Male , Membrane Proteins , Pregnancy , Pregnancy Outcome , Respiratory Distress Syndrome, Newborn/pathology , Superoxide Dismutase/metabolism , Thioredoxins/metabolism
20.
Circ Res ; 94(8): 1115-23, 2004 Apr 30.
Article in English | MEDLINE | ID: mdl-15016731

ABSTRACT

Endothelium-derived NO plays a critical role in the regulation of cardiovascular function and structure, as well as acting as a downstream mediator of the angiogenic response to numerous vascular growth factors. Although endothelial NO synthase (eNOS)-deficient mice are viable, minor congenital cardiac abnormalities have been reported and homozygous offspring exhibit high neonatal mortality out of proportion to the severity of these defects. The aim of the present report was to determine whether abnormalities of the pulmonary vascular development could contribute to high neonatal loss in eNOS-deficient animals. We now report that eNOS-deficient mice display major defects in lung morphogenesis, resulting in respiratory distress and death within the first hours of life in the majority of animals. Histological and molecular examination of preterm and newborn mutant lungs demonstrated marked thickening of saccular septae, with evidence of reduced surfactant material. Lungs of eNOS-deficient mice also exhibited a striking paucity of distal arteriolar branches and extensive regions of capillary hypoperfusion, together with misalignment of pulmonary veins, which represent the characteristic features of alveolar capillary dysplasia. We conclude that eNOS plays a previously unrecognized role in lung development, which may have relevance for clinical syndromes of neonatal respiratory distress.


Subject(s)
Lung/embryology , Nitric Oxide Synthase/deficiency , Pulmonary Alveoli/blood supply , Respiratory Distress Syndrome, Newborn/enzymology , Animals , Animals, Newborn , Apoptosis/drug effects , Basement Membrane/ultrastructure , Capillaries/pathology , Enzyme Induction , Enzyme Inhibitors/pharmacology , Extracellular Matrix/ultrastructure , Female , Fetal Proteins/deficiency , Fetal Proteins/physiology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Growth Substances/biosynthesis , Growth Substances/genetics , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Humans , Infant, Newborn , Litter Size/drug effects , Litter Size/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , NG-Nitroarginine Methyl Ester/pharmacology , NG-Nitroarginine Methyl Ester/toxicity , Neovascularization, Physiologic/genetics , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/physiology , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Obstetric Labor, Premature , Pregnancy , Pulmonary Surfactants/metabolism , Receptors, Growth Factor/biosynthesis , Receptors, Growth Factor/genetics , Respiratory Distress Syndrome, Newborn/embryology , Respiratory Distress Syndrome, Newborn/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...